1
|
Valdivia A, Vagadia PP, Guo G, O'Brien E, Matei D, Schiltz GE. Discovery and Characterization of PROTACs Targeting Tissue Transglutaminase (TG2). J Med Chem 2023. [PMID: 37449845 PMCID: PMC10388319 DOI: 10.1021/acs.jmedchem.2c01859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Tissue transglutaminase (TG2) is a multifunctional enzyme involved in the cross-linking of extracellular matrix proteins, formation of complexes with fibronectin (FN) and integrins, and GTP hydrolysis. TG2 is activated in several pathological conditions, including cancer. We recently described a novel series of ligands that bind to TG2 and inhibit its interaction with FN. Because TG2 acts via multiple mechanisms, we set out to pursue a targeted protein degradation strategy to abolish TG2's myriad functions. Here, we report the synthesis and characterization of a series of VHL-based degraders that reduce TG2 in ovarian cancer cells in a proteasome-dependent manner. Degradation of TG2 resulted in significantly reduced cancer cell adhesion and migration in vitro in scratch-wound and migration assays. These results strongly indicate that further development of more potent and in vivo efficient TG2 degraders could be a new strategy for reducing the dissemination of ovarian and other cancers.
Collapse
Affiliation(s)
- Andres Valdivia
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Purav P Vagadia
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Guangxu Guo
- WuXi AppTec, Shanghai 200131, People's Republic of China
| | - Eilidh O'Brien
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Jesse Brown VA Medical Center, Chicago, Illinois 60612, United States
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| |
Collapse
|
2
|
Suicide gene strategies applied in ovarian cancer studies. Cancer Gene Ther 2023:10.1038/s41417-023-00590-6. [PMID: 36717737 DOI: 10.1038/s41417-023-00590-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023]
Abstract
Ovarian cancer represents the most lethal gynecological malignancy among women in developed countries. Despite the recent innovations, the improvements in the 5-year survival rate have been insufficient and the management of this disease still remains a challenge. The fact that the majority of patients experience recurrent or resistant disease have substantiated the necessity of an innovative treatment. Among various strategies investigated, the recent strides made in gene delivery techniques have made gene therapy, including suicide gene strategies, a potential alternative for treating ovarian cancer. Various suicide gene candidates, which are capable of promoting cancer cell apoptosis directly after its entry or indirectly by prodrug administration, can be separated into three systems using enzyme-coding, toxin or pro-apoptotic genes. With this review, we aim to provide an overview of different suicide genes depending on therapeutic strategies, the vectors used to deliver these transgenes specifically to malignant cells, and the combined treatments of these genes with various therapeutic regimens.
Collapse
|
3
|
Sima LE, Matei D, Condello S. The Outside-In Journey of Tissue Transglutaminase in Cancer. Cells 2022; 11:cells11111779. [PMID: 35681474 PMCID: PMC9179582 DOI: 10.3390/cells11111779] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023] Open
Abstract
Tissue transglutaminase (TG2) is a member of the transglutaminase family that catalyzes Ca2+-dependent protein crosslinks and hydrolyzes guanosine 5′-triphosphate (GTP). The conformation and functions of TG2 are regulated by Ca2+ and GTP levels; the TG2 enzymatically active open conformation is modulated by high Ca2+ concentrations, while high intracellular GTP promotes the closed conformation, with inhibition of the TG-ase activity. TG2’s unique characteristics and its ubiquitous distribution in the intracellular compartment, coupled with its secretion in the extracellular matrix, contribute to modulate the functions of the protein. Its aberrant expression has been observed in several cancer types where it was linked to metastatic progression, resistance to chemotherapy, stemness, and worse clinical outcomes. The N-terminal domain of TG2 binds to the 42 kDa gelatin-binding domain of fibronectin with high affinity, facilitating the formation of a complex with β-integrins, essential for cellular adhesion to the matrix. This mechanism allows TG2 to interact with key matrix proteins and to regulate epithelial to mesenchymal transition and stemness. Here, we highlight the current knowledge on TG2 involvement in cancer, focusing on its roles translating extracellular cues into activation of oncogenic programs. Improved understanding of these mechanisms could lead to new therapeutic strategies targeting this multi-functional protein.
Collapse
Affiliation(s)
- Livia Elena Sima
- Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy, 060031 Bucharest, Romania;
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Salvatore Condello
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence:
| |
Collapse
|
4
|
Sima LE, Chen S, Cardenas H, Zhao G, Wang Y, Ivan C, Huang H, Zhang B, Matei D. Loss of host tissue transglutaminase boosts antitumor T cell immunity by altering STAT1/STAT3 phosphorylation in ovarian cancer. J Immunother Cancer 2021; 9:jitc-2021-002682. [PMID: 34593619 PMCID: PMC8487211 DOI: 10.1136/jitc-2021-002682] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tissue transglutaminase (TG2), an enzyme overexpressed in cancer cells, promotes metastasis and resistance to chemotherapy. Its distinct effects in cancer versus the host compartments have not been elucidated. METHODS Here, by using a TG2-/- syngeneic ovarian cancer mouse model, we assessed the effects of TG2 deficiency in the host tissues on antitumor immunity and tumor progression. Multicolor flow cytometry was used to phenotype immune cell populations in the peritoneal environment. Cancer cells recovered from malignant ascites were characterized by RNA sequencing, proliferation, and apoptosis assays. RESULTS We observed that host TG2 loss delayed tumor growth and ascites accumulation and caused increased infiltration of CD8+ T cells and decreased numbers of myeloid cells in the peritoneal fluid. Tumor antigen-specific CD8+ T cell cytotoxic responses were enhanced in ascites from TG2-/- versus TG2+/+ mice and CD8+ T cell depletion caused accelerated ascites accumulation in TG2-/- mice. CD8+ T cells from tumor-bearing TG2-/- mice displayed an effector T cell phenotype, differentiated toward effector memory (Tem). Mechanistically, absence of TG2 augmented signals promoting T cell activation, such as increased cytokine-induced STAT1 and attenuated STAT3 phosphorylation in T cells. Additionally, immune-suppressive myeloid cell populations were reduced in the peritoneal milieu of TG2-/- tumor-bearing mice. In response to the more robust immune response caused by loss of TG2, cancer cells growing intraperitoneally exhibited an interferon-γ(IFN-γ) responsive gene signature and underwent apoptosis. In human specimens, stromal, not tumor, TG2 expression correlated indirectly with numbers of tumor-infiltrating lymphocytes. CONCLUSIONS Collectively, our data demonstrate decreased tumor burden, increased activation and effector function of T cells, and loss of immunosuppressive signals in the tumor microenvironment of TG2-/- mice. We propose that TG2 acts as an attenuator of antitumor T cell immunity and is a new immunomodulatory target.
Collapse
Affiliation(s)
- Livia Elena Sima
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Siqi Chen
- Department of Medicine; Hematology/Oncology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Horacio Cardenas
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Guangyuan Zhao
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yinu Wang
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hao Huang
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bin Zhang
- Department of Medicine; Hematology/Oncology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
5
|
Yuan J, Lan H, Jiang X, Zeng D, Xiao S. Bcl‑2 family: Novel insight into individualized therapy for ovarian cancer (Review). Int J Mol Med 2020; 46:1255-1265. [PMID: 32945348 PMCID: PMC7447322 DOI: 10.3892/ijmm.2020.4689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022] Open
Abstract
Chemoresistance to platinum‑based chemotherapy for ovarian cancer in the advanced stage remains a formidable concern clinically. Increasing evidence has revealed that apoptosis represents the terminal events of the anti‑tumor mechanisms of a number of chemical drugs and has a close association with chemoresistance in ovarian cancer. The B‑cell lymphoma‑2 (Bcl‑2) family plays a crucial role in apoptosis and has a close association with chemoresistance in ovarian cancer. Some drugs that target Bcl‑2 family members have shown efficacy in overcoming the chemoresistance of ovarian cancer. A BH3 profiling assay was found to be able to predict how primed a cell is when treated with antitumor drugs. The present review summarizes the role of the Bcl‑2 family in mediating cell death in response to antitumor drugs and novel drugs that target Bcl‑2 family members. The application of the new functional assay, BH3 profiling, is also discussed herein. Furthermore, the present review presents the hypothesis that targeting Bcl‑2 family members may prove to be helpful for the individualized therapy of ovarian cancer in clinical practice and in laboratory research.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Hua Lan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaoyan Jiang
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Da Zeng
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
6
|
Xu J, Li Y, Hu H. Effects of lycopene on ovarian cancer cell line SKOV3 in vitro: Suppressed proliferation and enhanced apoptosis. Mol Cell Probes 2019; 46:101419. [PMID: 31279748 DOI: 10.1016/j.mcp.2019.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 11/26/2022]
Abstract
AIMS This study aims to investigate the influences of lycopene on ovarian cancer cells SKOV3 in vitro and the corresponding mechanism. METHODS SKOV3 cultures were divided into four groups and treated with lycopene at 0 (the control group), 1 × 10-6, 1 × 10-5 and 1 × 10-4 mol/L respectively. The proliferation rate of SKOV3 cells was determined using MTT colorimetric assay, and the apoptosis rate of SKOV3 cells was measured using flow cytometry. Western blotting was used to detect the expressions of Bax and Bcl-2. RESULTS When compared with the control group, the proliferation rates of SKOV3 cells in the three lycopene groups were significantly lower (P < 0.05) while the apoptosis rates were significantly higher (P < 0.05). After lycopene treatment, the protein expression of Bax (an apoptotic protein) in SKOV3 cells increased significantly, and the protein expression of Bcl-2 (an anti-apoptotic protein) decreased significantly. CONCLUSION Lycopene could inhibit the proliferation of SKOV3 cells and enhance their apoptosis in vitro. The induction of cell apoptosis might be mediated by up-regulating Bax expression and down-regulating Bcl-2 expression.
Collapse
Affiliation(s)
- Jian Xu
- Department of Gynaecology, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China
| | - Yuanfang Li
- Department of Reproduction, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China
| | - Hongyan Hu
- Department of Interventional Medicine, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China.
| |
Collapse
|
7
|
Zhu H, Chen L, Zhou W, Huang Z, Hu J, Dai S, Wang X, Huang X, He C. Over-expression of the ATP5J gene correlates with cell migration and 5-fluorouracil sensitivity in colorectal cancer. PLoS One 2013; 8:e76846. [PMID: 24124598 PMCID: PMC3790727 DOI: 10.1371/journal.pone.0076846] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 08/31/2013] [Indexed: 01/11/2023] Open
Abstract
Recently we found that ATP5J was over-expressed in tissue samples from patients with colorectal cancer. However, the clinical significance and function of the over-expression of ATP5J in these patients remains unclear. We investigated these issues in the current study. Our results indicated that expression of ATP5J was significantly higher in colorectal cancer tissue than in adjacent tissue, and it was also significantly higher in metastatic lymph nodes than in primary cancer tissue (P<0.05). A correlation between ATP5J expression and tumor differentiation was detected, but no correlation with gender, age, T stage, lymph node metastasis, or survival status was observed. Down-regulation of ATP5J expression attenuated the ability of cell migration and increased the sensitivity to 5-fluorouracil (5-Fu) in cells of the DLD1 cell line. Inversely, up-regulation of ATP5J expression enhanced cell migration and decreased 5-Fu sensitivity, suggesting that the function of ATP5J in colorectal cancer might involve cell migration and 5-Fu sensitivity.
Collapse
Affiliation(s)
- Hongbo Zhu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Biotherapy of Zhejiang province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Linlin Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Biotherapy of Zhejiang province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongting Huang
- Key Laboratory of Biotherapy of Zhejiang province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingzi Hu
- Department of Internal Medicine, Aviation Medical Evaluation and Training center of Airforce in Hangzhou, Hangzhou, China
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaowei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chao He
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail:
| |
Collapse
|
8
|
Brincks EL, Risk MC, Griffith TS. PMN and anti-tumor immunity--the case of bladder cancer immunotherapy. Semin Cancer Biol 2013; 23:183-9. [PMID: 23410637 DOI: 10.1016/j.semcancer.2013.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 01/21/2013] [Accepted: 02/01/2013] [Indexed: 12/01/2022]
Abstract
Urothelial carcinoma of the bladder accounts for ∼5% of all cancer deaths in humans. The majority of bladder tumors are non-muscle invasive at diagnosis, and there is a high rate of tumor recurrence and progression even after local surgical therapy. Thus, many patients require lifelong follow-up examinations that include additional prophylactic treatments in the event of recurrence. Since its first use in 1976, Mycobacterium bovis bacillus Calmette-Guerin (BCG) has been the treatment of choice for non-muscle invasive bladder cancer. Despite nearly 40 years of clinical use, the mechanism(s) by which intravesical administration of BCG results in elimination of bladder tumors remains undefined. Granulocytes (polymorphonuclear neutrophils (PMN)) are the predominant immune cell (in number) that enters the bladder after BCG installation, and a number of studies have highlighted the importance of PMN in the antitumor activity of BCG. Studies from our laboratory demonstrated presence of intracellular stores of the apoptosis-inducing protein TNF-related apoptosis-inducing ligand (TRAIL) in PMN that are rapidly released after interaction with BCG cell wall components, along with a correlation between increased urinary levels of TRAIL and BCG responsiveness. Mature PMN in circulation are terminally differentiated cells with limited biosynthetic capacity, so the proteins located in the distinct PMN granule populations are compartmentalized concomitant with their synthesis during myelopoiesis. Thus, understanding PMN production, localization, and release of TRAIL is important in the design of future BCG-based bladder tumor immunotherapy protocols.
Collapse
Affiliation(s)
- Erik L Brincks
- Department of Urology, University of Minnesota, Minneapolis, MN 55455, United States
| | | | | |
Collapse
|
9
|
Li XQ, Ke XZ, Wang YM. Treatment of malignant melanoma by downregulation of XIAP and overexpression of TRAIL with a conditionally replicating oncolytic adenovirus. Asian Pac J Cancer Prev 2013; 13:1471-6. [PMID: 22799350 DOI: 10.7314/apjcp.2012.13.4.1471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND AIM Currently available systemic therapies for malignant melanoma produce low response rates in patients, and more effective treatment modalities are clearly needed. The tumor necrosis factor (TNF)- related apoptosis-inducing ligand has a significant impact on therapy for patients with X-linked inhibitor of apoptosis protein-downregulation malignant melanoma. The primary objective of this study was to assess its therapeutic potential. MATERIALS AND METHODS We employed a conditionally replicating oncolytic adenoviral vector, named CRAd5.TRAIL/siXIAP, with the characteristics of over-expression of the therapeutic gene TRAIL and downregulation of XIAP in one vector. B16F10-luc cells were employed to detect anti-tumor activity of CRAd5.TRAIL/siXIAP in vitro and in vivo. RESULTS CRAd5.TRAIL/siXIAP enhanced caspase-8 activation and caspase-3 maturation in B16F10 cells in vitro. Furthermore, it more effectively infected and killed melanoma cells in vitro and in vivo than other adenoviruses. CONCLUSION Taken together, the combination of upregulation of TRAIL and downregulation of siXIAP with one oncolytic adenoviral vector holds promise for development of an effective therapy for melanomas and other common cancers.
Collapse
Affiliation(s)
- Xin-Qiu Li
- Department of Thyroid and Mammary Gland, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | | | | |
Collapse
|
10
|
Chai L, Liu S, Mao Q, Wang D, Li X, Zheng X, Xia H. A novel conditionally replicating adenoviral vector with dual expression of IL-24 and arresten inserted in E1 and the region between E4 and fiber for improved melanoma therapy. Cancer Gene Ther 2011; 19:247-54. [DOI: 10.1038/cgt.2011.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
11
|
Chen Y, Cao Y, Yang D, Li K, Wang Z, Zhu J, Bunjhoo H, Xiong S, Xu Y, Xiong W. Increase of the therapeutic effect on non-small-cell lung cancer cells with combination treatment of shRNA against Cyclin D1 and Bcl-xL in vitro. Exp Ther Med 2011; 3:255-260. [PMID: 22969878 DOI: 10.3892/etm.2011.381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 10/20/2011] [Indexed: 02/05/2023] Open
Abstract
Overexpression of Cyclin D1 and Bcl-xL proteins has often been found in non-small-cell lung cancer (NSCLC). These two genes may play a significant role in tumorigenesis. However, the combined inhibition of the two genes in vitro is unclear in NSCLC. In this study, the effect of a combined intervention on Cyclin D1 and Bcl-xL in NSCLC is assessed and discussed. Three recombinant plasmids that expressed a cytomegalovirus (CMV) promoter-driven micro30 short hairpin RNA (shRNA) targeting the Cyclin D1 gene (Cyclin D1 shRNA), the Bcl-xL gene (Bcl-xL shRNA) and a combination of the two genes (Cyclin D1-Bcl-xL shRNA), based on the plasmid pcDNA6.2-GW/EmGFP-miR, were constructed. The cell lines A549 and NCI-H441 were divided into four groups; blank control (untreated cells), Cyclin D1 shRNA, Bcl-xL shRNA and Cyclin D1-Bcl-xL shRNA (transfected cells), respectively. The expression of mRNA and protein of Cyclin D1 or Bcl-xL was detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting, respectively. The apoptosis and proliferation of the two cell lines were evaluated by dimethylthiazol-diphenyltetrazolium bromide (MTT), cell count and flow cytometry. The recombinant plasmid sufficiently mediated the RNA interference (RNAi) effects in A549 and NCI-H441 cells. The expression levels of mRNA and protein of Cyclin D1 or Bcl-xL in the three intervention groups were significantly reduced compared to the untreated cells (P<0.05). No statistical differences were found among the combined shRNAs and single shRNA regarding Cyclin D1 or Bcl-xL, respectively (P>0.05). In the assessment of proliferation and apoptosis, it was found that in all three intervention groups there was significant inhibition of cell proliferation and promotion of cell apoptosis compared with the untreated cells (P<0.05). Furthermore, the combined interference of the two genes was more effective than either single interference (P<0.05). Our results suggested that the combined targeting of Cyclin D1 and Bcl-xL genes has potential for NSCLC investigation, providing increased efficacy over Cyclin D1 or Bcl-xL inhibition alone.
Collapse
Affiliation(s)
- Ying Chen
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zu L, Liu H, Chen J, Zhou Q. [Current status and prospect of lung cancer gene therapy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:758-62. [PMID: 21924046 PMCID: PMC5999612 DOI: 10.3779/j.issn.1009-3419.2011.09.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | | | | | | |
Collapse
|
13
|
Norian LA, James BR, Griffith TS. Advances in Viral Vector-Based TRAIL Gene Therapy for Cancer. Cancers (Basel) 2011; 3:603-20. [PMID: 24212631 PMCID: PMC3756379 DOI: 10.3390/cancers3010603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 01/28/2011] [Accepted: 01/30/2011] [Indexed: 12/16/2022] Open
Abstract
Numerous biologic approaches are being investigated as anti-cancer therapies in an attempt to induce tumor regression while circumventing the toxic side effects associated with standard chemo- or radiotherapies. Among these, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has shown particular promise in pre-clinical and early clinical trials, due to its preferential ability to induce apoptotic cell death in cancer cells and its minimal toxicity. One limitation of TRAIL use is the fact that many tumor types display an inherent resistance to TRAIL-induced apoptosis. To circumvent this problem, researchers have explored a number of strategies to optimize TRAIL delivery and to improve its efficacy via co-administration with other anti-cancer agents. In this review, we will focus on TRAIL-based gene therapy approaches for the treatment of malignancies. We will discuss the main viral vectors that are being used for TRAIL gene therapy and the strategies that are currently being attempted to improve the efficacy of TRAIL as an anti-cancer therapeutic.
Collapse
Affiliation(s)
- Lyse A. Norian
- Department of Urology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
| | - Britnie R. James
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
| | - Thomas S. Griffith
- Department of Urology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-319-335-7581; Fax: +1-319-353-4556
| |
Collapse
|
14
|
Barbolina MV, Adley BP, Kelly DL, Fought AJ, Scholtens D, Shea LD, Sharon Stack M. Motility-related actinin alpha-4 is associated with advanced and metastatic ovarian carcinoma. J Transl Med 2008; 88:602-14. [PMID: 18362906 PMCID: PMC2849305 DOI: 10.1038/labinvest.2008.25] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Advanced and metastatic ovarian cancer is a leading cause of death from gynecologic malignancies. A more detailed understanding of the factors controlling invasion and metastasis may lead to novel anti-metastatic therapies. To model cellular interactions that occur during intraperitoneal metastasis, comparative cDNA microarray analysis and confirmatory real-time reverse transcription PCR (RT-PCR) were employed to uncover changes in gene expression that may occur in late stage ovarian cancer in response to microenvironmental cues, particularly native three-dimensional collagen I. Gene expression in human ovarian carcinoma tissues was evaluated on the RNA and protein level using real-time RT-PCR and immunohistochemistry. Cell invasion and migration were evaluated in a collagen invasion assay and a scratch wound assay. Three-dimensional collagen I culture led to differential expression of several genes. The role of actinin alpha-4 (ACTN4), a cytoskeleton-associated protein implicated in the regulation of cell motility, was examined in detail. ACTN4 RNA and protein expression were associated with advanced and metastatic human ovarian carcinoma. This report demonstrates that a cytoskeletal-associated protein ACTN4 is upregulated by three-dimensional collagen culture conditions, leading to increased invasion and motility of ovarian cancer cells. Expression of ACTN4 in human ovarian tumors was found to be associated with advanced-stage disease and peritoneal metastases.
Collapse
Affiliation(s)
- Maria V. Barbolina
- Department of Chemical & Biochemical Engineering, Northwestern University, Chicago, IL 60611
| | - Brian P. Adley
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - David L. Kelly
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Angela J. Fought
- Department of Preventive Medicine, Northwestern University, Chicago, IL 60611
| | - Denise Scholtens
- Department of Preventive Medicine, Northwestern University, Chicago, IL 60611
| | - Lonnie D. Shea
- Department of Chemical & Biochemical Engineering, Northwestern University, Chicago, IL 60611
| | - M. Sharon Stack
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO 65212,To whom correspondence and reprint requests should be addressed: M. Sharon Stack, Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, M214E Medical Sciences Bldg, Columbia, MO 65212, Ph. 573-884-7301,
| |
Collapse
|
15
|
Zhu H, Zhu Y, Hu J, Hu W, Liao Y, Zhang J, Wang D, Huang X, Fang B, He C. Adenovirus-mediated small hairpin RNA targeting Bcl-XL as therapy for colon cancer. Int J Cancer 2007; 121:1366-72. [PMID: 17534896 DOI: 10.1002/ijc.22856] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bcl-XL, an anti-apoptotic protein of Bcl-2 family, is overexpressed in colon cancers. To determine Bcl-XL's potential feasibility as a therapeutic target, we constructed a recombinant adenovirus that expressed a U6 promoter-driven small hairpin RNA (shRNA) targeting Bcl-XL (Ad/Bcl-XL shRNA) and evaluated the vector's ability to induce RNA interference in vivo and alter apoptosis induction in colon cancer cells and tumours. Ad/Bcl-XL shRNA effectively knocked down Bcl-XL expression in colon cancer cells and decreased their viability. Treatment with Ad/Bcl-XL shRNA but not control vectors led to dramatically increased cleavage of cellular apoptosis-related enzymes caspase-9, caspase-3 and poly(ADP-ribose) polymerase. Ad/Bcl-XL shRNA also significantly suppressed the growth of subcutaneous tumours derived from DLD1 cells in a nude mouse model and did so without causing any obvious damage to normal tissues or normal human fibroblasts. Together, our results support the feasibility of using adenovirus-mediated RNA interference therapy targeting Bcl-XL against colon cancers and warrant further studies of its safety and efficacy.
Collapse
Affiliation(s)
- Hongbo Zhu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kendrick JE, Estes JM, Straughn JM, Alvarez RD, Buchsbaum DJ. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its therapeutic potential in breast and gynecologic cancers. Gynecol Oncol 2007; 106:614-21. [PMID: 17602728 DOI: 10.1016/j.ygyno.2007.05.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 05/30/2007] [Accepted: 05/30/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The relationship between the apoptotic pathway and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising area of scientific interest for cancer researchers. TRAIL-receptor-activating agents have demonstrated favorable in vitro and in vivo activity for the treatment of several malignancies including breast and gynecologic cancers. METHODS This article reviews the available peer-reviewed literature and our own institution's experience with specific TRAIL-receptor-activating agents. Emphasis was placed on the apoptotic/TRAIL mechanism, preclinical evaluation, and phase I studies in various malignancies. RESULTS Preclinical and early phase I studies indicate that these novel agents are safe with enhanced target specificity for malignancy. When these targeted agents are combined with conventional chemotherapy drugs or radiation therapy, they appear to increase cell death over single-agent modalities. CONCLUSIONS TRAIL-receptor-activating agents represent an exciting new class of targeted therapies that hold promise to improve the treatment of women with breast and gynecologic malignancies.
Collapse
Affiliation(s)
- James E Kendrick
- Division of Gynecologic Oncology, The University of Alabama at Birmingham, 619 19th Street South, OHB 538, Birmingham, AL 35249, USA.
| | | | | | | | | |
Collapse
|
17
|
Abstract
Lung cancer patients suffer a 15% overall survival despite advances in chemotherapy, radiation therapy, and surgery due to the usual finding of advanced disease at diagnosis. Attempts to improve survival in advanced disease using various combinations of chemotherapy have demonstrated that no regimen is superior, suggesting a therapeutic plateau and the need for novel, more specific, and less toxic therapeutic strategies. Techniques have been developed that allow transfer of functional genes into mammalian cells, such as those that block activated tumor-promoting oncogenes and/or those that replace inactivated tumor-suppressing or apoptosis-promoting genes. This article will discuss the therapeutic implications of these molecular changes associated with bronchogenic carcinomas, and will then review the status of gene therapies for treatment of lung cancer.
Collapse
Affiliation(s)
- Eric M Toloza
- Duke Thoracic Oncology Program, Duke University Medical Center, Box 3048, Durham, NC 27710, USA.
| |
Collapse
|
18
|
Abstract
Lung cancer patients suffer a 15% overall survival despite advances in chemotherapy, radiation therapy, and surgery. This unacceptably low survival rate is due to the usual finding of advanced disease at diagnosis. However, multimodality strategies using conventional therapies only minimally improve survival rates even in early stages of lung cancer. Attempts to improve survival in advanced disease using various combinations of platinum-based chemotherapy have demonstrated that no regimen is superior, suggesting a therapeutic plateau and the need for novel, more specific, and less toxic therapeutic strategies. Over the past three decades, the genetic etiology of cancer has been gradually delineated, albeit not yet completely. Understanding the molecular events that occur during the multistep process of bronchogenic carcinogenesis may make these tasks more surmountable. During these same three decades, techniques have been developed which allow transfer of functional genes into mammalian cells. For example, blockade of activated tumor-promoting oncogenes or replacement of inactivated tumor-suppressing or apoptosis-promoting genes can be achieved by gene therapy. This article will discuss the therapeutic implications of these molecular changes associated with bronchogenic carcinomas and will then review the status of gene therapies for treatment of lung cancer.
Collapse
Affiliation(s)
- Eric M Toloza
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
19
|
|
20
|
Cordelier P, Bienvenu C, Lulka H, Marrache F, Bouisson M, Openheim A, Strayer DS, Vaysse N, Pradayrol L, Buscail L. Replication-deficient rSV40 mediate pancreatic gene transfer and long-term inhibition of tumor growth. Cancer Gene Ther 2006; 14:19-29. [PMID: 16990845 DOI: 10.1038/sj.cgt.7700987] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Pancreatic cancer is one of the most aggressive and devastating human malignancies. There is an urgent need for more effective therapy for patients with advanced disease. In this context, genetic therapy potentially represents a rational new approach to treating pancreatic cancer, which could provide an adjunct to conventional options. Because of the promise of recombinant SV40 vectors, we tested their ability to deliver a transgene, and to target a transcript, so as to inhibit pancreatic tumors growth in vivo. BxPC3 and Capan-1 cells were efficiently transduced using SV40 vectors without selection, as compared to synthetic vectors PEI. SV40 vectors were as efficient as adenoviral vectors, and provided long-term transgene expression. Next, we devised a SV40-derived, targeted gene therapy approach of pancreatic cancer, by combining hTR tumor-specific promoter with sst2 somatostatin receptor tumor-suppressor gene. In vitro cell proliferation was strongly impaired following administration of SV(hTR-sst2). SV40-derived sst2-mediated antiproliferative effect was dependent on the local production of somatostatin. In vivo, intratumoral gene transfer of sst2 using rSV40 vectors resulted in a marked inhibition of Capan-1 tumor progression, and proliferation. These results represent the initial steps toward a novel approach to the gene therapy of pancreatic cancer using SV40 as a vector.
Collapse
Affiliation(s)
- P Cordelier
- INSERM U531, Institut Louis Bugnard, IFR31, Toulouse, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhao L, Dong A, Gu J, Liu Z, Zhang Y, Zhang W, Wang Y, He L, Qian C, Qian Q, Liu X. The antitumor activity of TRAIL and IL-24 with replicating oncolytic adenovirus in colorectal cancer. Cancer Gene Ther 2006; 13:1011-22. [PMID: 16799468 DOI: 10.1038/sj.cgt.7700969] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melanoma differentiation associated gene-7 (Mda-7)/IL-24 was previously cloned into ZD55 (an adenovirus with E1B55 deleted) to form ZD55-IL-24, which had much better antitumor effect than Ad-IL-24. According to its good antitumor properties, ZD55-IL-24 has been used in preclinical studies. But ZD55-IL-24 alone still could not completely eradicate established tumors in all nude mice. It was reported that IL-24 could induce and enhance the activity of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) (a member of tumor necrosis factor (TNF) superfamily). Accordingly, the combined use of ZD55-IL-24 and ZD55-TRAIL was carried out in this study. Treatment with both ZD55-IL-24 and ZD55-TRAIL could induce more significant apoptosis in cancer cells in vitro compared with ZD55-IL-24 or ZD55-TRAIL alone. The combination of the two replicative adenoviruses had better antitumor activity in vivo than that of single oncolytic adenovirus and led to complete eradication of xenograft tumors in all treated mice. Upregulation of TRAIL was observed in tumor cells infected with ZD55-IL-24 and studies of the apoptotic cascade regulators indicate that ZD55-IL-24 could further enhance the activation of apoptosis through the TNF family of death receptors. We demonstrated for the first time the potential therapeutic effect of combined ZD55-IL-24 with ZD55-TRAIL for the targeted therapy of cancer.
Collapse
Affiliation(s)
- L Zhao
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, The Graduate School, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Liu P, Mao H, Hou P. Synergistic antitumor effect of tumor necrosis factor-related apoptosis-inducing ligand combined with cisplatin in ovarian carcinoma cell lines in vitro and in vivo. Int J Gynecol Cancer 2006; 16:538-48. [PMID: 16681723 DOI: 10.1111/j.1525-1438.2006.00507.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to exert selectively cytotoxic activity against many tumor cells but not normal cells. In this study, we evaluated the antitumor activity of TRAIL and cisplatin (CDDP) both separately and combined in the human ovarian cancer cell lines. In vitro study showed that TRAIL elicited significant cell apoptosis of cell lines 3AO, SKOV3, and OVCAR3 in a dose- and time-dependent manner (P < 0.05), while normal ovarian epithelial cells were resistant; this toxicity-free effect may be the result of upregulation of TRAIL receptors DcR1 and DcR2. Combined TRAIL and CDDP therapy produced more profound cell killing in 3AO cells than each alone (P < 0.05), and CDDP could upregulate the expression of both death and decoy TRAIL receptors. To further evaluate the apoptosis-inducing effects of TRAIL and the combination therapy, the abdominally and subcutaneously spread tumors in nude mice via inoculation of 3AO cells were established, and treatment of TRAIL resulted in a dose- and time-dependent inhibition of tumor growth while slight damage was observed in normal tissues. Furthermore, combined TRAIL and CDDP therapy had a synergistic effect in the regression of established ovarian cancer xenografts than TRAIL treatment alone (P < 0.05). We also examined the apoptosis-related gene expression in the transplantation tumors after TRAIL treatment, and the data suggested that the intracellular mechanism of TRAIL may be associated with downregulation of Bcl-2 and upregulation of CD95 and Apo2.7.
Collapse
Affiliation(s)
- P Liu
- Department of Obstetrics and Gynecology, QiLu Hospital of ShanDong University, Jinan, ShanDong, China
| | | | | |
Collapse
|
23
|
Choi E, Kim Y, Kim K. The combination of TRAIL treatment and cancer cell selective expression of TRAIL-death receptor DR4 induces cell death in TRAIL-resistant cancer cells. Yonsei Med J 2006; 47:55-62. [PMID: 16502485 PMCID: PMC2687581 DOI: 10.3349/ymj.2006.47.1.55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The human telomerase reverse transcriptase (hTERT) promoter can be used for the tumor-specific expression of transgenes in order to induce selective cancer cell death. The hTERT core promoter is active in cancer cells but not in normal cells. To examine whether the combination of TNF-related apoptosis inducing ligand (TRAIL) treatment and cancer cell-selective expression of the TRAIL-death receptor could induce cell death in TRAIL-resistant cancer cells, we generated a death receptor-4 (DR4)-expressing adenovirus (Ad-hTERT-DR4), in which the expression of DR4 is driven by the hTERT promoter. Upon infection, DR4 expression was slightly increased in cancer cell lines, and cell death was observed in TRAIL-resistant cancer cell lines but not in normal human cells when DR4 infection was combined with TRAIL treatment. We also generated an adenovirus that expresses a secretable isoleucine zipper (ILZ)-fused, extracellular portion of TRAIL (Ad-ILZ- TRAIL). In cells infected with Ad-ILZ-TRAIL, TRAIL was expressed, secreted, oligomerized and biologically active in the induction of apoptosis in TRAIL-sensitive cancer cells. When Ad-hTERT-DR4 infected TRAIL-resistant HCE4 cells and Ad-ILZ-TRAIL infected TRAIL-resistant HCE7 cells were co-cultured, cell deaths were evident 24 h after co-culture. Taken together, our results reveal that the combination of TRAIL and cancer cell-specific expression of DR4 has the potential to overcome the resistance of cancer cells to TRAIL without inducing significant cell death in normal cells.
Collapse
Affiliation(s)
- Eunah Choi
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Brain Korea 21 Project for Medical Science of Yonsei University, Seoul, Korea
| | - Youngtae Kim
- Department of Gynecology and Obstetrics, Yonsei University College of Medicine, Brain Korea 21 Project for Medical Science of Yonsei University, Seoul, Korea
| | - Kunhong Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Brain Korea 21 Project for Medical Science of Yonsei University, Seoul, Korea
| |
Collapse
|
24
|
Jacob D, Schumacher G, Bahra M, Davis J, Zhu HB, Zhang LD, Teraishi F, Neuhaus P, Fang BL. Fiber-modified adenoviral vector expressing the tumor necrosis factor-related apoptosis-inducing ligand gene from the human telomerase reverse transcriptase promoter induces apoptosis in human hepatocellular carcinoma cells. World J Gastroenterol 2005; 11:2552-6. [PMID: 15849810 PMCID: PMC4305742 DOI: 10.3748/wjg.v11.i17.2552] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Because of a major resistance to chemotherapy, prognosis of hepatocellular carcinoma (HCC) is still poor. New treatments are required and gene therapy may be an option. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in multiple malignant tumors, and using adenoviral vectors has shown a targeted tumor-specific therapy. However, repeated administration of adenoviral vectors can lead to cell resistance, which may be caused by the initial coxsackie-adenovirus receptor (CAR). One technique to overcome resistance is the use of modified adenoviral vectors containing an Arg-Gly-Asp (RGD) sequence. In this study we constructed an adenoviral vector (designated Ad/TRAIL-F/RGD) with RGD-modified fibers, expressing the TRAIL gene from the human telomerase reverse transcriptase (hTERT) promoter, and evaluated its antitumor activity in HCC cell lines.
METHODS: To investigate the effects of Ad/TRAIL-F/RGD in human HCC cell lines Hep G2 and Hep 3b, cells were infected with Ad/CMV-GFP (vector control), Ad/gTRAIL (positive control), and Ad/TRAIL-F/RGD. Phosphate-buffered saline (PBS) was used as control. Cell viability was determined by proliferation assay (XTT), and apoptosis induction by fluorescence activated cell sorting (FACS).
RESULTS: Cells treated with Ad/TRAIL-F/RGD and Ad/gTRAIL showed a significantly reduced cell viability in comparison to PBS and Ad/CMV-GFP treatment in both cell lines. Whereas, treatment with PBS and Ad/CMV-GFP had no cell-killing effect. The reduced cell viability was caused by induction of apoptosis as shown by FACS analysis. The amount of apoptotic cells was similar after incubation with Ad/gTRAIL and Ad/TRAIL-F/RGD.
CONCLUSION: The new RGD modified vector Ad/TRAIL-F/RGD could become a potent therapeutic agent for the treatment of HCC, adenovirus resistant tumors, and CAR low or negative cancer cells.
Collapse
Affiliation(s)
- Dietmar Jacob
- Department of General, Visceral and Transplantation Surgery, Humboldt University of Berlin, CharitA Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Freytag SO, Kim JH, Brown SL, Barton K, Lu M, Chung M. Gene therapy strategies to improve the effectiveness of cancer radiotherapy. Expert Opin Biol Ther 2005; 4:1757-70. [PMID: 15500404 DOI: 10.1517/14712598.4.11.1757] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Having the ability to alter the genetic makeup of a cancer cell by gene transfer is a potentially powerful strategy for treating human cancer. However, a low efficiency of gene delivery in vivo and poor tumour specificity has prevented the widespread implementation of this technology in the clinic. Despite these formidable obstacles, the first successful application of gene therapy in the treatment of cancer may occur when it is combined with local modalities such as radiation therapy. A small number of gene therapy strategies have been evaluated in clinical trials in combination with external beam radiation therapy. The combined therapy has been well-tolerated and has not exacerbated the side effects of radiation therapy. Gene transfer and tumour cell destruction has been demonstrated in vivo. Although the results await confirmation in larger, prospective Phase III trials, there is suggestive evidence that the combined therapies may be demonstrating better than expected antitumour activity. Our vast knowledge of the molecular defects that drive the cancer process, coupled with our expanding understanding of the genes responsible for tumour cell radioresistance, have spawned the development of rational, targeted gene therapies designed to increase tumour cell radiosensitivity. Here, the results of the clinical trials conducted so far will be reviewed, followed by a description of new approaches under development at present.
Collapse
Affiliation(s)
- Svend O Freytag
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI 48202-3405, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Rodina AV, Sladkova LV, Obuchova VV, Vezirkhanova TZ, Moskaleva EY, Prusakova OV, Beletskii IP, Belushkina NN, Strelnikov VV, Ivanov MA, Severin SE, Severin ES. Inactivation and sensitization of tumor cells after transfection with gene Bax. Mol Biol 2005. [DOI: 10.1007/s11008-005-0005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Zhu H, Zhang L, Huang X, Davis JJ, Jacob DA, Teraishi F, Chiao P, Fang B. Overcoming acquired resistance to TRAIL by chemotherapeutic agents and calpain inhibitor I through distinct mechanisms. Mol Ther 2004; 9:666-73. [PMID: 15120327 DOI: 10.1016/j.ymthe.2004.02.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Accepted: 02/06/2004] [Indexed: 11/24/2022] Open
Abstract
We recently found that repeated application of adenovectors expressing the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or recombinant TRAIL proteins to TRAIL-susceptible cancer cells resulted in selection and expansion of TRAIL-resistant cells. Overcoming this acquired resistance to TRAIL is desirable for TRAIL-mediated cancer therapy. Here we demonstrate that several chemotherapeutic agents, including 5-fluorouracil (5-FU) and mitomycin, and calpain inhibitor I, an NFkappaB inhibitor, can overcome acquired resistance to TRAIL in DLD1 colon cancer cells. The combination of TRAIL (approved gene symbol TNFSF10) gene therapy and 5-FU enhanced tumor suppression in vivo in nude mice bearing subcutaneous tumors established from TRAIL-resistant colon cancer cells. Whereas treatment with the combination of TRAIL and 5-FU or mitomycin led to enhanced activation of caspase-3, the combination of TRAIL and calpain inhibitor I resulted in enhanced activation of both caspase-8 and caspase-3. Moreover, mitomycin, but not 5-FU or calpain inhibitor I, induced overexpression of the BAX gene, which was correlated with enhanced TRAIL-induced cell killing in TRAIL-resistant DLD1 cells. Together, these results suggest that acquired resistance to TRAIL can be overcome by different mechanisms and that combinations of TRAIL gene therapy and chemotherapy may be a useful approach for cancer treatment.
Collapse
Affiliation(s)
- Hongbo Zhu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kaliberov SA, Kaliberova LN, Stockard CR, Grizzle WE, Buchsbaum DJ. Adenovirus-mediated FLT1-targeted proapoptotic gene therapy of human prostate cancer. Mol Ther 2004; 10:1059-70. [PMID: 15564138 DOI: 10.1016/j.ymthe.2004.08.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2004] [Accepted: 08/30/2004] [Indexed: 12/19/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is of particular interest in the development of prostate carcinoma therapeutics as it preferentially induces apoptosis of tumor cells. To employ adenoviral vectors for highly efficient and specific TRAIL gene transfer into cancer cells could overcome some potential problems for recombinant TRAIL. The vascular endothelial growth factor receptor FLT-1 is involved in regulation of angiogenesis and tumor growth, invasion, and metastasis of prostate carcinoma. FLT-1 expression is observed in both tumor endothelial cells and prostate cancer cells. We developed an adenoviral vector encoding the TRAIL gene under control of the FLT1 promoter (AdFlt-TRAIL), which produced endothelial and prostate cancer cell death. The combination of ionizing radiation and adenovirus-driven TRAIL expression overcame human prostate cancer cell resistance to TRAIL. Furthermore, in vivo administration of AdFlt-TRAIL at the site of tumor growth in combination with radiation treatment produced significant suppression of the growth of DU145 human prostate tumor xenografts in athymic nude mice. Our results suggest that specific TRAIL delivery employing the FLT1 promoter can effectively inhibit tumor growth and demonstrate the advantage of combination radiotherapy and gene therapy for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Sergey A Kaliberov
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
29
|
Sova P, Ren XW, Ni S, Bernt KM, Mi J, Kiviat N, Lieber A. A tumor-targeted and conditionally replicating oncolytic adenovirus vector expressing TRAIL for treatment of liver metastases. Mol Ther 2004; 9:496-509. [PMID: 15093180 DOI: 10.1016/j.ymthe.2003.12.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2003] [Accepted: 12/17/2003] [Indexed: 01/09/2023] Open
Abstract
We have constructed a new capsid-modified adenovirus (Ad) vector that specifically replicates in tumor cells and expresses TNF-related apoptosis-inducing ligand (TRAIL). The Ad capsid contains short-shafted fibers derived from Ad serotype 35, which allow for efficient infection of malignant tumor cells, and largely avoids innate toxicity after intravenous application. Replication-dependent homologous recombination in Ad genomes was used to achieve tumor-specific expression of Ad E1a (to mediate viral replication) and TRAIL (to mediate apoptosis and enhance release of progeny virus from infected cells). We demonstrated that our oncolytic vector (Ad5/35.IR-E1A/TRAIL) induced apoptosis in human tumor cell lines derived from colorectal, lung, prostate, and liver cancer. Both in vitro and in vivo tumor models showed efficient intratumoral spread of this vector. In a model for metastatic colon cancer, tail vein infusion of Ad5/35.IR-E1A/TRAIL resulted in elimination of preestablished liver metastases. Intravenous injection of this vector caused a transient elevation of serum glutamic pyruvic transaminase in tumor-bearing mice, which we attributed to factors released from apoptotic tumor cells. Liver histology analyzed at day 14 after virus injection did not show signs of hepatocellular damage. This new oncolytic vector represents a potentially efficient means for gene therapy of metastatic cancer.
Collapse
Affiliation(s)
- Pavel Sova
- Department of Pathology, University of Washington, Box 357720, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abou El Hassan MAI, Mastenbroek DCJ, Gerritsen WR, Giaccone G, Kruyt FAE. Overexpression of Bcl2 abrogates chemo- and radiotherapy-induced sensitisation of NCI-H460 non-small-cell lung cancer cells to adenovirus-mediated expression of full-length TRAIL. Br J Cancer 2004; 91:171-7. [PMID: 15173860 PMCID: PMC2364740 DOI: 10.1038/sj.bjc.6601910] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo-2L) is a promising novel anticancer agent that selectively induces apoptosis in tumour cells and the activity of which can be enhanced by combined treatment with chemo- or radiotherapy. For therapeutic purposes, the use of full-length TRAIL may be favourable to recombinant TRAIL based on its increased tumour cell killing potential, and the delivery of TRAIL at the tumour site by adenovirus vectors may provide an approach to overcome the short half-life of recombinant TRAIL and hepatocyte toxicity in vivo. Here, we constructed an adenoviral vector expressing full-length TRAIL (AdTRAIL) and studied the potential of chemo- and radiotherapy in enhancing AdTRAIL-induced apoptosis in non-small cell lung cancer (NSCLC) H460 cells and normal cells and, in addition, investigated the mechanism of AdTRAIL-induced apoptosis. AdTRAIL effectively killed H460 cells, which we previously showed to have a deficiency in mitochondria-dependent apoptosis by downstream activation of caspase-8 rather than caspase-9. Further analyses revealed that AdTRAIL induces death receptor- and mitochondria-dependent apoptosis that could be partially suppressed by Bcl2 overexpression. Combined treatment with doxorubicin (DOX), cisplatin (CDDP), paclitaxel (PTX) and radiation strongly enhanced AdTRAIL-induced cytotoxicity in a synergistic way. Synergy was accompanied by the cleavage of Bid and an increase in caspase-8 processing that was abolished by Bcl2 overexpression, indicating that the Bid-mitochondrial amplification loop is functional in H460 cells. Moreover, combination treatment did not alter the tumour selectivity of AdTRAIL since normal human fibroblasts (NHFs) remained resistant under these conditions. These findings further indicate that the combined use of chemo/radiotherapy and adenovirus-produced full-length TRAIL may provide a valuable treatment option for NSCLC.
Collapse
Affiliation(s)
- M A I Abou El Hassan
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - D C J Mastenbroek
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - W R Gerritsen
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - G Giaccone
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - F A E Kruyt
- Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Medical Oncology, Room Br 232, PO Box 7057, 1007 MB, Amsterdam, The Netherlands. E-mail:
| |
Collapse
|
31
|
Groot-Wassink T, Aboagye EO, Wang Y, Lemoine NR, Keith WN, Vassaux G. Noninvasive Imaging of the Transcriptional Activities of Human Telomerase Promoter Fragments in Mice. Cancer Res 2004; 64:4906-11. [PMID: 15256462 DOI: 10.1158/0008-5472.can-04-0426] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have assessed the feasibility of positron emission tomography (PET) and ex vivo gamma-counting to measure the pattern of expression of telomerase promoter fragments in vivo. Promoter fragments from either the RNA [human telomerase RNA (hTR)] or the catalytic components [human telomerase reverse transcriptase (hTERT)] of the telomerase genes were used to drive the expression of the sodium iodide symporter PET reporter gene in recombinant adenoviruses. Both promoter fragments provided cancer-selective expression that could be visualized and quantitated by PET. The transcriptional activity of the hTR promoter was found to be consistently stronger than that of the hTERT promoter. Both promoters appear therefore to be good candidates for safe use in gene therapy, and PET imaging can be used to assess the selectivity of promoters in vivo. Given that this methodology is directly scalable to humans, imaging gene expression using the sodium iodide symporter PET reporter gene could be applied to measure telomerase promoter activity in humans.
Collapse
Affiliation(s)
- Thomas Groot-Wassink
- Cancer Research United Kingdom Molecular Oncology Unit, Department of Imaging Sciences, Faculty of Medicine, Imperial College, Hammersmith Campus, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Odoux C, Albers A. Additive Effects of TRAIL and Paclitaxel on Cancer Cells: Implications for Advances in Cancer Therapy. VITAMINS AND HORMONES 2004; 67:385-407. [PMID: 15110187 DOI: 10.1016/s0083-6729(04)67020-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
In cancer therapy outgrowth of chemoresistant tumor cells is the most important factor that ultimately determines-apart from immediate adverse effects during treatment-the life span and prognosis of cancer patients. Despite many advances in cancer treatment and the integration of supportive medications, including new and better drugs for pain management, antiemesis, infection, and reconstitution of the hematopoietic system, both toxic effects and the development of resistance in response to the treatment remain a major problem. New treatment regimens have to be developed to target cancer more specifically using multiple cellular pathways. This will reduce toxic effects as well as the development of chemoresistance. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is the ligand for death receptors that belong to the TNF death receptor family. TRAIL triggers apoptosis in vitro in various cancer cell types. The antitumor drug, Paclitaxel (PA) was shown to increase the survival of patients with cancer. In in vitro experiments, PA also induces apoptosis in cancer cells. Together, PA and TRAIL lead to tumor regression in in vivo therapy and induce apoptosis through the interaction of TNF family death receptors, caspase activation, and?or cytochrome c release from mitochondria. PA and TRAIL complement each other using two distinct pathways that trigger apoptosis in addition to the anti-microtubule effect of PA. The combination of TRAIL and PA suppresses tumor growth that is otherwise resistant to treatment with either PA or TRAIL alone, by improving proapoptotic effects of the drugs. This observation support the use of the PA and TRAIL in future clinical trials.
Collapse
Affiliation(s)
- Christine Odoux
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pennsylvania, USA
| | | |
Collapse
|
33
|
Lin T, Zhang L, Davis J, Gu J, Nishizaki M, Ji L, Roth JA, Xiong M, Fang B. Combination of TRAIL gene therapy and chemotherapy enhances antitumor and antimetastasis effects in chemosensitive and chemoresistant breast cancers. Mol Ther 2003; 8:441-8. [PMID: 12946317 DOI: 10.1016/s1525-0016(03)00203-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
We recently found that breast cancer cell lines that are resistant to chemotherapy or to the recombinant TRAIL protein are susceptible to TRAIL gene therapy. However, it is unclear whether a combination of TRAIL gene therapy and chemotherapy will have enhanced antitumor activity or can be used for the treatment of metastasis. In this study, we investigated the combined effect of TRAIL gene therapy and chemotherapeutic agents, including doxorubicin, paclitaxel, vinorelbine, gemcitabine, irinotecan, and floxuridine, in different breast cancer cell lines. In all the cell lines tested, including a breast cancer cell line that is resistant to chemotherapy, the combination of TRAIL gene therapy and cytotoxic agents had either a synergistic or an additive effect. An in vivo study showed that aerosolized administration of an adenovector expressing the GFP-TRAIL fusion protein from the human telomerase reverse transcriptase promoter (designated Ad/gTRAIL) also decreased the number of lung metastases from both doxorubicin-sensitive and doxorubicin-resistant breast cancer cell lines. The combination of TRAIL gene therapy and chemotherapy resulted in a further reduction of lung metastatic nodules with minimal toxicity. These results suggest that a combination of TRAIL gene therapy and chemotherapy is effective in the treatment of metastatic diseases.
Collapse
Affiliation(s)
- Tongyu Lin
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, 77030, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Nicosia SV, Bai W, Cheng JQ, Coppola D, Kruk PA. Oncogenic pathways implicated in ovarian epithelial cancer. Hematol Oncol Clin North Am 2003; 17:927-43. [PMID: 12959183 DOI: 10.1016/s0889-8588(03)00056-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Characterization of intracellular signaling pathways should lead to a better understanding of ovarian epithelial carcinogenesis and provide an opportunity to interfere with signal transduction targets involved in ovarian tumor cell growth, survival, and progression. Challenges toward such an effort are significant because many of these signals are part of cascades within an intricate and likely redundant intracellular signaling network (Fig.1). For instance, a given signal may activate a dual intracellular pathway (ie, MEK1-MAPK and PI3K/Akt required for fibronectin-dependent activation of matrix metalloproteinase 9). A single pathway also may transduce more than one biologic or oncogenic signal (ie, PI3K signaling in epithelial and endothelial cell growth and sprouting of neovessels). Despite these challenges, evidence for therapeutic targeting of signal transduction pathways is accumulating in human cancer. For instance, the EGF-specific tyrosine kinase inhibitor ZD 1839 (Iressa) may have a beneficial therapeutic effect on ovarian epithelial cancer. Therapy of this cancer may include inhibitors of PI kinase (quercetin), ezrin and PIP kinase (genistein). The G protein-coupled family of receptors, including LPA, also is an attractive target to drugs, although their frequent pleiotropic functions may be at times toxic and lack specificity. Because of the lack of notable toxicity, PI3K/Akt pathway inhibitors such as FTIs are a promising targeted therapy of ovarian epithelial cancer. Increasing insight into the oncogenic pathways involved in ovarian epithelial cancer also is helping clinicians to understand better the phenomenon of chemoresistance in this malignancy. Oncogenic activation of gamma-synuclein promotes cell survival and provides resistance to paclitaxel, but such a resistance is partially overcome by an MEK inhibitor that suppresses ERK activity. Ovarian epithelial cancer is a complex group of neoplasms with an overall poor prognosis. Comprehension of this cancer pathobiology suffers because of an incomplete understanding of precursor lesions and the absence of an orthotopic animal model until very recently. It can be predicted with confidence, however, that the discovery of potent inhibitors of signal transduction and the development of discovery tools, such as proteomics and metabolomics, may change the way by which clinicians may now address basic biomedical questions in this insidious and lethal disease.
Collapse
Affiliation(s)
- Santo V Nicosia
- Department of Pathology and Laboratory Medicine, University of South Florida College of Medicine, 12901 Bruce B. Downs Boulevard, MDC Box 11, Tampa, FL 33612, USA.
| | | | | | | | | |
Collapse
|
35
|
|